Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Mais filtros

Tipo de documento
Intervalo de ano de publicação
1.
Epilepsy Behav ; 146: 109363, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37499576

RESUMO

Lysine, as an essential amino acid, predominantly undergoes metabolic processes through the saccharopine pathway, whereas a smaller fraction follows the pipecolic acid pathway. Although the liver is considered the primary organ for lysine metabolism, it is worth noting that lysine catabolism also takes place in other tissues and organs throughout the body, including the brain. Enzyme deficiency caused by pathogenic variants in its metabolic pathway may lead to a series of neurometabolic diseases, among which glutaric aciduria type 1 and pyridoxine-dependent epilepsy have the most significant clinical manifestations. At present, through research, we have a deeper understanding of the multiple pathophysiological mechanisms related to these diseases, including intracerebral accumulation of neurotoxic metabolites, imbalance between GABAergic and glutamatergic neurotransmission, energy deprivation due to metabolites, and the dysfunction of antiquitin. Because of the complexity of these diseases, their clinical manifestations are also diverse. The early implementation of lysine-restricted diets and supplementation with arginine and carnitine has reported positive impacts on the neurodevelopmental outcomes of patients. Presently, there is more robust evidence supporting the effectiveness of these treatments in glutaric aciduria type 1 compared with pyridoxine-dependent epilepsy.


Assuntos
Encefalopatias Metabólicas , Epilepsia , Humanos , Lisina/metabolismo , Epilepsia/metabolismo , Encefalopatias Metabólicas/complicações , Encefalopatias Metabólicas/metabolismo
2.
Mol Genet Metab ; 131(3): 325-340, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-33069577

RESUMO

Glutaric acidemia type 1 (GA1) is a disorder of cerebral organic acid metabolism resulting from biallelic mutations of GCDH. Without treatment, GA1 causes striatal degeneration in >80% of affected children before two years of age. We analyzed clinical, biochemical, and developmental outcomes for 168 genotypically diverse GA1 patients managed at a single center over 31 years, here separated into three treatment cohorts: children in Cohort I (n = 60; DOB 2006-2019) were identified by newborn screening (NBS) and treated prospectively using a standardized protocol that included a lysine-free, arginine-enriched metabolic formula, enteral l-carnitine (100 mg/kg•day), and emergency intravenous (IV) infusions of dextrose, saline, and l-carnitine during illnesses; children in Cohort II (n = 57; DOB 1989-2018) were identified by NBS and treated with natural protein restriction (1.0-1.3 g/kg•day) and emergency IV infusions; children in Cohort III (n = 51; DOB 1973-2016) did not receive NBS or special diet. The incidence of striatal degeneration in Cohorts I, II, and III was 7%, 47%, and 90%, respectively (p < .0001). No neurologic injuries occurred after 19 months of age. Among uninjured children followed prospectively from birth (Cohort I), measures of growth, nutritional sufficiency, motor development, and cognitive function were normal. Adherence to metabolic formula and l-carnitine supplementation in Cohort I declined to 12% and 32%, respectively, by age 7 years. Cessation of strict dietary therapy altered plasma amino acid and carnitine concentrations but resulted in no serious adverse outcomes. In conclusion, neonatal diagnosis of GA1 coupled to management with lysine-free, arginine-enriched metabolic formula and emergency IV infusions during the first two years of life is safe and effective, preventing more than 90% of striatal injuries while supporting normal growth and psychomotor development. The need for dietary interventions and emergency IV therapies beyond early childhood is uncertain.


Assuntos
Erros Inatos do Metabolismo dos Aminoácidos/genética , Encefalopatias Metabólicas/genética , Encéfalo/metabolismo , Corpo Estriado/metabolismo , Glutaril-CoA Desidrogenase/deficiência , Glutaril-CoA Desidrogenase/genética , Erros Inatos do Metabolismo dos Aminoácidos/dietoterapia , Erros Inatos do Metabolismo dos Aminoácidos/epidemiologia , Erros Inatos do Metabolismo dos Aminoácidos/metabolismo , Encéfalo/patologia , Encefalopatias Metabólicas/dietoterapia , Encefalopatias Metabólicas/epidemiologia , Encefalopatias Metabólicas/metabolismo , Carnitina/metabolismo , Criança , Pré-Escolar , Corpo Estriado/patologia , Dieta , Feminino , Glutaril-CoA Desidrogenase/metabolismo , Humanos , Lactente , Recém-Nascido , Lisina/metabolismo , Masculino
3.
J Nutr ; 150(Suppl 1): 2556S-2560S, 2020 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-33000154

RESUMO

Lysine is an essential amino acid, and inherited diseases of its metabolism therefore represent defects of lysine catabolism. Although some of these enzyme defects are not well described yet, glutaric aciduria type I (GA1) and antiquitin (2-aminoadipic-6-semialdehyde dehydrogenase) deficiency represent the most well-characterized diseases. GA1 is an autosomal recessive disorder due to a deficiency of glutaryl-CoA dehydrogenase. Untreated patients exhibit early onset macrocephaly and may present a neurological deterioration with regression and movement disorder at the time of a presumably "benign" infection most often during the first year of life. This is associated with a characteristic neuroimaging pattern with frontotemporal atrophy and striatal injuries. Diagnosis relies on the identification of glutaric and 3-hydroxyglutaric acid in urine along with plasma glutarylcarnitine. Treatment consists of a low-lysine diet aiming at reducing the putatively neurotoxic glutaric and 3-hydroxyglutaric acids. Additional therapeutic measures include administration of l-carnitine associated with emergency measures at the time of intercurrent illnesses aiming at preventing brain injury. Early treated (ideally through newborn screening) patients exhibit a favorable long-term neurocognitive outcome, whereas late-treated or untreated patients may present severe neurocognitive irreversible disabilities. Antiquitin deficiency is the most common form of pyridoxine-dependent epilepsy. α-Aminoadipic acid semialdehyde (AASA) and Δ-1-piperideine-6-carboxylate (P6C) accumulate proximal to the enzymatic block. P6C forms a complex with pyridoxal phosphate (PLP), a key vitamer of pyridoxine, thereby reducing PLP bioavailability and subsequently causing epilepsy. Urinary AASA is a biomarker of antiquitin deficiency. Despite seizure control, only 25% of the pyridoxine-treated patients show normal neurodevelopment. Low-lysine diet and arginine supplementation are proposed in some patients with decrease of AASA, but the impact on neurodevelopment is unclear. In summary, GA1 and antiquitin deficiency are the 2 main human defects of lysine catabolism. Both include neurological impairment. Lysine dietary restriction is a key therapy for GA1, whereas its benefits in antiquitin deficiency appear less clear.


Assuntos
Aldeído Desidrogenase/deficiência , Erros Inatos do Metabolismo dos Aminoácidos/metabolismo , Encefalopatias Metabólicas Congênitas/metabolismo , Encefalopatias Metabólicas/metabolismo , Encéfalo/metabolismo , Epilepsia/metabolismo , Glutaril-CoA Desidrogenase/deficiência , Lisina/metabolismo , Ácido 2-Aminoadípico/análogos & derivados , Ácido 2-Aminoadípico/metabolismo , Aldeído Desidrogenase/metabolismo , Erros Inatos do Metabolismo dos Aminoácidos/terapia , Arginina/uso terapêutico , Encéfalo/patologia , Encefalopatias Metabólicas/terapia , Encefalopatias Metabólicas Congênitas/terapia , Carnitina/análogos & derivados , Carnitina/metabolismo , Carnitina/uso terapêutico , Epilepsia/terapia , Glutaratos/metabolismo , Glutaril-CoA Desidrogenase/metabolismo , Humanos , Doenças Metabólicas/genética , Doenças Metabólicas/metabolismo , Doenças Metabólicas/terapia , Fosfato de Piridoxal/metabolismo , Piridoxina/metabolismo , Piridoxina/uso terapêutico
4.
Int J Mol Sci ; 21(19)2020 Sep 25.
Artigo em Inglês | MEDLINE | ID: mdl-32992790

RESUMO

Riboflavin is the biological precursor of two important flavin cofactors-flavin adenine dinucleotide (FAD) and flavin mononucleotide (FMN)-that are critical prosthetic groups in several redox enzymes. While dietary supplementation with riboflavin is a recognized support therapy in several inborn errors of metabolism, it has yet unproven benefits in several other pathologies affecting flavoproteins. This is the case for glutaric aciduria type I (GA-I), a rare neurometabolic disorder associated with mutations in the GCDH gene, which encodes for glutaryl-coenzyme A (CoA) dehydrogenase (GCDH). Although there are a few reported clinical cases that have responded to riboflavin intake, there is still not enough molecular evidence supporting therapeutic recommendation. Hence, it is necessary to elucidate the molecular basis in favor of riboflavin supplementation in GA-I patients. Here, using a combination of biochemical and biophysical methodologies, we investigate the clinical variant GCDH-p.Val400Met as a model for a phenotype associated with severe deflavinylation. Through a systematic analysis, we establish that recombinant human GCDH-p.Val400Met is expressed in a nonfunctional apo form, which is mainly monomeric rather than tetrameric. However, we show that exogenous FAD is a driver for structural reorganization of the mutant enzyme with concomitant functional recovery, improved thermolability, and resistance to trypsin digestion. Overall, these results establish proof of principle for the beneficial effects of riboflavin supplementation in GA-I patients.


Assuntos
Erros Inatos do Metabolismo dos Aminoácidos , Encefalopatias Metabólicas , Glutaril-CoA Desidrogenase/deficiência , Glutaril-CoA Desidrogenase/genética , Riboflavina , Erros Inatos do Metabolismo dos Aminoácidos/metabolismo , Encefalopatias Metabólicas/metabolismo , Glutaril-CoA Desidrogenase/química , Glutaril-CoA Desidrogenase/efeitos dos fármacos , Glutaril-CoA Desidrogenase/metabolismo , Humanos , Mutação , Dobramento de Proteína/efeitos dos fármacos , Estabilidade Proteica/efeitos dos fármacos , Proteínas Recombinantes , Riboflavina/farmacologia
5.
Biochim Biophys Acta Mol Basis Dis ; 1865(9): 2420-2427, 2019 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-31181292

RESUMO

The deficiency of the enzyme glutaryl-CoA dehydrogenase leads to predominant accumulation of glutaric acid (GA) in the organism and is known as glutaric acidemia type I (GA1). Despite the mechanisms of brain damage involved in GA1 are not fully understood, oxidative stress may be involved in this process. Treatment is based on protein/lysine (Lys) restriction and l-carnitine (L-car) supplementation. L-car was recently shown to have an important antioxidant role. A knockout mice model (Gcdh-/-) submitted to a dietary overload of Lys was developed to better understand the GA1 pathogenesis. In this study, we evaluated L-car and glutarylcarnitine levels, the lipid and protein damage, reactive oxygen species (ROS) production and antioxidant enzymes activities in striatum of Gcdh-/- and wild-type (WT) mice. We also determined the effect of the L-car treatment on these parameters. Thirty-day-old Gcdh-/- and WT mice were fed a normal chow (0.9% Lys) or submitted to a high Lys diet (4.7%) for 72 h. Additionally, these animals were administered with three intraperitoneal injections of saline or L-car in different times. Gcdh-/- mice were deficient in L-car and presented a higher glutarylcarnitine levels. They also presented lipid and protein damage, an increased ROS production and altered antioxidant enzymes compared to WT mice. Additionally, mice exposed to Lys overload presented higher alterations in these parameters than mice under normal diet, which were significantly decreased or normalized in those receiving L-car. Thus, we demonstrated a new beneficial effect of the L-car treatment attenuating or abolishing the oxidative stress process in Gcdh-/- mice.


Assuntos
Carnitina/farmacologia , Corpo Estriado/metabolismo , Glutaril-CoA Desidrogenase/genética , Lisina/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Erros Inatos do Metabolismo dos Aminoácidos/metabolismo , Erros Inatos do Metabolismo dos Aminoácidos/patologia , Erros Inatos do Metabolismo dos Aminoácidos/veterinária , Animais , Encefalopatias Metabólicas/metabolismo , Encefalopatias Metabólicas/patologia , Encefalopatias Metabólicas/veterinária , Carnitina/análogos & derivados , Carnitina/metabolismo , Dieta/veterinária , Modelos Animais de Doenças , Glutaril-CoA Desidrogenase/deficiência , Glutaril-CoA Desidrogenase/metabolismo , Glutationa Peroxidase/metabolismo , Lisina/sangue , Camundongos , Camundongos Knockout , Espécies Reativas de Oxigênio/metabolismo , Superóxido Dismutase/metabolismo
6.
Am Soc Clin Oncol Educ Book ; 38: 822-828, 2018 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-30231327

RESUMO

There are 15.5 million cancer survivors in the United States because of, in part, improvements in therapy. As a result, there will be an increased burden of long- and late-term complications of cancer care, such as metabolic alterations. These metabolic changes will include alterations in bone resorption, obesity, hypercholesterolemia, and diabetes mellitus. The majority of cancer treatment-related toxicities have focused on endocrine therapy; however, chemotherapy and supportive medications, such as steroids, contribute to the development of these disorders. Because of the chronicity of these metabolic changes and their impact on morbidity, cancer risk, and outcomes as well other negative effects, including musculoskeletal pain and vasomotor symptoms, alternative strategies must be developed. These strategies should include nonpharmacologic approaches. Here, we summarize metabolic changes secondary to cancer care and integrative approaches to help alleviate therapy-associated toxicities.


Assuntos
Terapia Combinada/efeitos adversos , Neoplasias/complicações , Antagonistas de Androgênios/efeitos adversos , Antagonistas de Androgênios/uso terapêutico , Antineoplásicos Hormonais/efeitos adversos , Antineoplásicos Hormonais/uso terapêutico , Encefalopatias Metabólicas/etiologia , Encefalopatias Metabólicas/metabolismo , Encefalopatias Metabólicas/patologia , Terapia Combinada/métodos , Prestação Integrada de Cuidados de Saúde , Gerenciamento Clínico , Glucocorticoides/efeitos adversos , Glucocorticoides/uso terapêutico , Humanos , Hipogonadismo/complicações , Síndrome Metabólica/etiologia , Neoplasias/metabolismo , Neoplasias/terapia , Osteoporose/etiologia
7.
J Cell Biochem ; 119(12): 10021-10032, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30129250

RESUMO

The deficiency of the enzyme glutaryl-CoA dehydrogenase, known as glutaric acidemia type I (GA-I), leads to the accumulation of glutaric acid (GA) and glutarilcarnitine (C5DC) in the tissues and body fluids, unleashing important neurotoxic effects. l-carnitine (l-car) is recommended for the treatment of GA-I, aiming to induce the excretion of toxic metabolites. l-car has also demonstrated an important role as antioxidant and anti-inflammatory in some neurometabolic diseases. This study evaluated GA-I patients at diagnosis moment and treated the oxidative damage to lipids, proteins, and the inflammatory profile, as well as in vivo and in vitro DNA damage, reactive nitrogen species (RNS), and antioxidant capacity, verifying if the actual treatment with l-car (100 mg kg-1 day-1 ) is able to protect the organism against these processes. Significant increases of GA and C5DC were observed in GA-I patients. A deficiency of carnitine in patients before the supplementation was found. GA-I patients presented significantly increased levels of isoprostanes, di-tyrosine, urinary oxidized guanine species, and the RNS, as well as a reduced antioxidant capacity. The l-car supplementation induced beneficial effects reducing these biomarkers levels and increasing the antioxidant capacity. GA, in three different concentrations, significantly induced DNA damage in vitro, and the l-car was able to prevent this damage. Significant increases of pro-inflammatory cytokines IL-6, IL-8, GM-CSF, and TNF-α were shown in patients. Thus, the beneficial effects of l-car presented in the treatment of GA-I are due not only by increasing the excretion of accumulated toxic metabolites, but also by preventing oxidative damage.


Assuntos
Erros Inatos do Metabolismo dos Aminoácidos/metabolismo , Encefalopatias Metabólicas/metabolismo , Carnitina/farmacologia , Dano ao DNA , Glutaril-CoA Desidrogenase/deficiência , Estresse Oxidativo , Antioxidantes/farmacologia , Antioxidantes/uso terapêutico , Carnitina/uso terapêutico , Criança , Pré-Escolar , Feminino , Glutaril-CoA Desidrogenase/efeitos dos fármacos , Glutaril-CoA Desidrogenase/metabolismo , Humanos , Lactente , Masculino , Substâncias Protetoras/farmacologia , Substâncias Protetoras/uso terapêutico , Espécies Reativas de Nitrogênio
8.
Biochim Biophys Acta Mol Basis Dis ; 1864(1): 263-273, 2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-29107091

RESUMO

Alzheimer's disease (AD) is an amyloid-related neurodegenerative disorder and is also considered to be a metabolic disease. Thus, investigation of metabolic mechanisms of amyloid pathology progression is of substantial importance for the diagnosis, prevention and treatment of AD. In the present study, cognitive function and brain metabolism were explored in the transgenic APP/PS1 mouse model of amyloid pathology at different ages. Using an NMR-based metabolomic approach, we examined metabolic changes in six different brain regions of wild-type and APP/PS1 mice at 1, 5 and 10months of age. Learning and memory performance in mice was evaluated using the Morris water maze test. Furthermore, a generalized linear mixed model was employed to analyze the interaction effect between the mouse-type and brain region (or age) on metabolic alterations. Brain region-specific changes in energy metabolism occurred prior to a very early-stage of amyloid pathology (1month of age) in APP/PS1 mice. A hypermetabolic state was identified in the brains of APP/PS1 mice at 5months of age, and the hypothalamus was identified as the main brain region that underwent significant metabolic alterations. The cognitive function of APP/PS1 mice was impaired at 10months of age; moreover, the hypermetabolic state identified in various brain regions at 5months of age was also significantly decreased. In conclusion, our results suggest that a hypothalamic metabolism abnormality may comprise a potential indicator for the early-diagnosis and monitoring of amyloid pathology progression.


Assuntos
Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Precursor de Proteína beta-Amiloide/genética , Hipotálamo/metabolismo , Hipotálamo/patologia , Presenilina-1/genética , Doença de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Amiloidose/genética , Amiloidose/metabolismo , Amiloidose/patologia , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Encefalopatias Metabólicas/genética , Encefalopatias Metabólicas/metabolismo , Encefalopatias Metabólicas/patologia , Cognição/fisiologia , Modelos Animais de Doenças , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Presenilina-1/metabolismo
9.
J Inherit Metab Dis ; 40(1): 75-101, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27853989

RESUMO

Glutaric aciduria type I (GA-I; synonym, glutaric acidemia type I) is a rare inherited metabolic disease caused by deficiency of glutaryl-CoA dehydrogenase located in the catabolic pathways of L-lysine, L-hydroxylysine, and L-tryptophan. The enzymatic defect results in elevated concentrations of glutaric acid, 3-hydroxyglutaric acid, glutaconic acid, and glutaryl carnitine in body tissues, which can be reliably detected by gas chromatography/mass spectrometry (organic acids) and tandem mass spectrometry (acylcarnitines). Most untreated individuals with GA-I experience acute encephalopathic crises during the first 6 years of life that are triggered by infectious diseases, febrile reaction to vaccinations, and surgery. These crises result in striatal injury and consequent dystonic movement disorder; thus, significant mortality and morbidity results. In some patients, neurologic disease may also develop without clinically apparent crises at any age. Neonatal screening for GA-I us being used in a growing number of countries worldwide and is cost effective. Metabolic treatment, consisting of low lysine diet, carnitine supplementation, and intensified emergency treatment during catabolism, is effective treatment and improves neurologic outcome in those individuals diagnosed early; treatment after symptom onset, however, is less effective. Dietary treatment is relaxed after age 6 years and should be supervised by specialized metabolic centers. The major aim of this second revision of proposed recommendations is to re-evaluate the previous recommendations (Kölker et al. J Inherit Metab Dis 30:5-22, 2007b; J Inherit Metab Dis 34:677-694, 2011) and add new research findings, relevant clinical aspects, and the perspective of affected individuals.


Assuntos
Erros Inatos do Metabolismo dos Aminoácidos/diagnóstico , Erros Inatos do Metabolismo dos Aminoácidos/tratamento farmacológico , Encefalopatias Metabólicas/diagnóstico , Encefalopatias Metabólicas/tratamento farmacológico , Glutaril-CoA Desidrogenase/deficiência , Erros Inatos do Metabolismo dos Aminoácidos/metabolismo , Encefalopatias Metabólicas/metabolismo , Suplementos Nutricionais , Glutaratos/metabolismo , Glutaril-CoA Desidrogenase/metabolismo , Humanos , Lisina/metabolismo
10.
Neuroscience ; 308: 64-74, 2015 Nov 12.
Artigo em Inglês | MEDLINE | ID: mdl-26343296

RESUMO

The brain of children affected by organic acidemias develop acute neurodegeneration linked to accumulation of endogenous toxic metabolites like glutaric (GA), 3-hydroxyglutaric (3-OHGA), methylmalonic (MMA) and propionic (PA) acids. Excitotoxic and oxidative events are involved in the toxic patterns elicited by these organic acids, although their single actions cannot explain the extent of brain damage observed in organic acidemias. The characterization of co-adjuvant factors involved in the magnification of early toxic processes evoked by these metabolites is essential to infer their actions in the human brain. Alterations in the kynurenine pathway (KP) - a metabolic route devoted to degrade tryptophan to form NAD(+) - produce increased levels of the excitotoxic metabolite quinolinic acid (QUIN), which has been involved in neurodegenerative disorders. Herein we investigated the effects of subtoxic concentrations of GA, 3-OHGA, MMA and PA, either alone or in combination with QUIN, on early toxic endpoints in rat brain synaptosomes. To establish specific mechanisms, we pre-incubated synaptosomes with different protective agents, including the endogenous N-methyl-d-aspartate (NMDA) receptor antagonist kynurenic acid (KA), the antioxidant S-allylcysteine (SAC) and the nitric oxide synthase (NOS) inhibitor nitro-l-arginine methyl ester (l-NAME). While the incubation of synaptosomes with toxic metabolites at subtoxic concentrations produced no effects, their co-incubation (QUIN+GA, +3-OHGA, +MMA or +PA) decreased the mitochondrial function and increased reactive oxygen species (ROS) formation and lipid peroxidation. For all cases, this effect was partially prevented by KA and l-NAME, and completely avoided by SAC. These findings suggest that early damaging events elicited by organic acids involved in metabolic acidemias can be magnified by toxic synergism with QUIN, and this process is mostly mediated by oxidative stress, and in a lesser extent by excitotoxicity and nitrosative stress. Therefore, QUIN can be hypothesized to contribute to the pathophysiology of brain degeneration in children with metabolic acidemias.


Assuntos
Erros Inatos do Metabolismo dos Aminoácidos/metabolismo , Encefalopatias Metabólicas/metabolismo , Encéfalo/metabolismo , Glutaratos/metabolismo , Glutaril-CoA Desidrogenase/deficiência , Ácido Quinolínico/metabolismo , Sinaptossomos/metabolismo , Animais , Encéfalo/efeitos dos fármacos , Modelos Animais de Doenças , Glutaratos/toxicidade , Glutaril-CoA Desidrogenase/metabolismo , Peroxidação de Lipídeos/efeitos dos fármacos , Masculino , Ácido Metilmalônico/metabolismo , Ácido Metilmalônico/toxicidade , Fármacos Neuroprotetores/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Estresse Oxidativo/fisiologia , Propionatos/metabolismo , Propionatos/toxicidade , Ácido Quinolínico/toxicidade , Ratos Wistar , Espécies Reativas de Oxigênio/metabolismo , Sinaptossomos/efeitos dos fármacos
11.
PLoS One ; 9(10): e110181, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25333616

RESUMO

Acute neurological crises involving striatal degeneration induced by a deficiency of glutaryl-CoA dehydrogenase (GCDH) and the accumulation of glutaric (GA) and 3-hydroxyglutaric acid (3-OHGA) are considered to be the most striking features of glutaric aciduria type I (GA1). In the present study, we investigated the mechanisms of apoptosis and energy metabolism impairment in our novel GA1 neuronal model. We also explored the effects of appropriate amounts of amino acids (2 mM arginine, 2 mM homoarginine, 0.45 g/L tyrosine and 10 mM leucine) and 2 g/L glucose on these cells. Our results revealed that the novel GA1 neuronal model effectively simulates the hypermetabolic state of GA1. We found that leucine, tyrosine, arginine, homoarginine or glucose treatment of the GA1 model cells reduced the gene expression of caspase-3, caspase-8, caspase-9, bax, fos, and jun and restored the intracellular NADH and ATP levels. Tyrosine, arginine or homoarginine treatment in particular showed anti-apoptotic effects; increased α-ketoglutarate dehydrogenase complex (OGDC), fumarase (FH), and citrate synthase (CS) expression; and relieved the observed impairment in energy metabolism. To the best of our knowledge, this study is the first to investigate the protective mechanisms of amino acids and glucose in GA1 at the cellular level from the point of view of apoptosis and energy metabolism. Our data support the results of previous studies, indicating that supplementation of arginine and homoarginine as a dietary control strategy can have a therapeutic effect on GA1. All of these findings facilitate the understanding of cell apoptosis and energy metabolism impairment in GA1 and reveal new therapeutic perspectives for this disease.


Assuntos
Erros Inatos do Metabolismo dos Aminoácidos/metabolismo , Aminoácidos/metabolismo , Encefalopatias Metabólicas/metabolismo , Técnicas de Cultura de Células , Glucose/metabolismo , Glutaril-CoA Desidrogenase/deficiência , Trifosfato de Adenosina/metabolismo , Erros Inatos do Metabolismo dos Aminoácidos/genética , Animais , Animais Recém-Nascidos , Apoptose/genética , Proteínas Reguladoras de Apoptose/genética , Proteínas Reguladoras de Apoptose/metabolismo , Encefalopatias Metabólicas/genética , Sobrevivência Celular , Células Cultivadas , Ciclo do Ácido Cítrico/genética , Corpo Estriado/citologia , Citometria de Fluxo , Expressão Gênica , Glutaril-CoA Desidrogenase/genética , Glutaril-CoA Desidrogenase/metabolismo , NAD/metabolismo , Neurônios/metabolismo , Ratos
12.
J Med Food ; 16(7): 577-86, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23875897

RESUMO

Diabetic encephalopathy (DE) is one of the severe complications in patients with diabetes mellitus. Paeonol, an active compound isolated from the root bark of Paeonia suffruticosa, has significant antidiabetic activity in vivo. However, its underlying beneficial effects on DE were unclear. In the present study, the protective activity of paeonol on DE was evaluated in streptozotocin (STZ)-induced diabetic rats. Paeonol at 50 and 100 mg/kg significantly increased body weight and decreased blood glucose levels, glycosylated serum proteins, and serum advanced glycation end products (AGEs) levels. Immunohistochemistry assays and Western blot analysis revealed a significant decrease in expressions on receptor for advanced glycation end products (RAGE) and nuclear factor kappa B (NF-κB) in hippocampus and cerebral cortical neurons after paeonol treatment. Furthermore, paeonol significantly increased glutathione content and remarkedly decreased induced nitric oxide synthase activity in hippocampus tissue. Our findings indicated that paeonol could improve the pathological damage of DE in STZ-induced diabetic rats. It might be associated with the modulating AGEs/RAGE/NF-κB pathway. This study suggested that paeonol might be a promising candidate for the prevention and treatment of DE.


Assuntos
Antioxidantes/administração & dosagem , Encefalopatias Metabólicas/tratamento farmacológico , Diabetes Mellitus Experimental/complicações , Diabetes Mellitus Tipo 2/complicações , Hipoglicemiantes/administração & dosagem , Paeonia/química , Animais , Encefalopatias Metabólicas/etiologia , Encefalopatias Metabólicas/genética , Encefalopatias Metabólicas/metabolismo , Produtos Finais de Glicação Avançada/metabolismo , Humanos , Masculino , NF-kappa B/genética , NF-kappa B/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Estreptozocina
13.
Exp Gerontol ; 48(4): 414-21, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23419687

RESUMO

Aging is associated with decreased insulin sensitivity and impaired cerebral glucose homeostasis. These changes increase neural sensitivity to metabolic damage contributing to cognitive decline, being the decrease in plasma estrogen following menopause one of the main factors involved in aged females. Phytoestrogens as genistein are structurally similar to 17ß-estradiol, bind to estrogen receptors, and can evoke both estrogenic and anti-estrogenic effects. Estrogens and phytoestrogens have neuroprotective potential, but the physiological mechanisms are not fully understood. Young and aged female Wistar rats were ovariectomized and treated acutely with 17ß-estradiol (1.4µg/kg body weight), genistein (10 or 40 mg/kg body weight), or vehicle. Cortical expression of glucose transporter-3 (GLUT-3) and -4 (GLUT-4), cytochrome c oxidase (CO), estrogen receptor-α (ERα) and -ß (ERß) was measured by Western blotting. There was an age-related decline in GLUT-4, CO and ERß levels. Both drugs, estradiol and genistein, were able to reverse GLUT-3 downregulation in the cortex following late ovariectomy. However, genistein was the only treatment able to restore completely GLUT-4 levels in aged rats. In contrast, estradiol was more potent than genistein at increasing CO, a marker of cerebral oxidative metabolism. As regards ER levels, estradiol increased the ERα67 quantity diminished by late ovariectomy, while genistein did the same with the other ERα isoform, ERα46, highlighting drug-specific differences in expression changes for both isoforms. On the other hand, no treatment-related differences were found regarding ERß levels. Therefore, genistein like estradiol could be suitable treatments against cortical metabolic dysfunction caused by aging. These treatments may hold promise as neuroprotective strategies against diabetes and age-related neurodegenerative diseases.


Assuntos
Encefalopatias Metabólicas , Córtex Cerebral/metabolismo , Estradiol , Genisteína , Redes e Vias Metabólicas/efeitos dos fármacos , Ovariectomia/efeitos adversos , Envelhecimento/metabolismo , Envelhecimento/psicologia , Animais , Encefalopatias Metabólicas/tratamento farmacológico , Encefalopatias Metabólicas/etiologia , Encefalopatias Metabólicas/metabolismo , Cognição/efeitos dos fármacos , Cognição/fisiologia , Estradiol/metabolismo , Estradiol/farmacologia , Estrogênios/metabolismo , Estrogênios/farmacologia , Feminino , Genisteína/metabolismo , Genisteína/farmacologia , Glucose/metabolismo , Humanos , Menopausa/metabolismo , Modelos Animais , Fármacos Neuroprotetores/metabolismo , Fármacos Neuroprotetores/farmacologia , Oxirredução/efeitos dos fármacos , Fitoestrógenos/metabolismo , Fitoestrógenos/farmacologia , Ratos , Ratos Wistar , Receptores de Estrogênio/metabolismo , Resultado do Tratamento
14.
J Inherit Metab Dis ; 36(3): 525-33, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-22971958

RESUMO

BACKGROUND: Metabolic treatment in glutaric aciduria type I (GA-I) including a low lysine diet with lysine-free, tryptophan-reduced amino acid supplements (AAS), carnitine supplementation and early start of emergency treatment during putatively threatening episodes of intermittent febrile illness dramatically improves the outcome and thus has been recommended by an international guideline group (Kölker et al, J Inherit Metab Dis 30:5-22, 2007). However, possible affection of linear growth, weight gain and biochemical follow-up monitoring has not been studied systematically. METHODS: Thirty-three patients (n = 29 asymptomatic, n = 4 dystonic) with GA-I who have been identified by newborn screening in Germany from 1999 to 2009 were followed prospectively during the first six years of life. Dietary treatment protocols, anthropometrical and biochemical parameters were longitudinally evaluated. RESULTS: Mean daily intake as percentage of guideline recommendations was excellent for lysine (asymptomatic patients: 101 %; dystonic patients: 103 %), lysine-free, tryptophan-reduced AAS (108 %; 104 %), energy (106 %; 110 %), and carnitine (92 %; 102 %). Low lysine diet did not affect weight gain (mean SDS 0.05) but mildly impaired linear growth in asymptomatic patients (mean SDS -0.38), while dystonic patients showed significantly reduced weight gain (mean SDS -1.32) and a tendency towards linear growth retardation (mean SDS -1.03). Patients treated in accordance with recent recommendations did not show relevant abnormalities of routine biochemical follow-up parameters. INTERPRETATION: Low lysine diet promotes sufficient intake of essential nutrients and anthropometric development in asymptomatic children up to age 6 year, whereas individualized nutritional concepts are required for dystonic patients. Revised recommendations for biochemical monitoring might be required for asymptomatic patients.


Assuntos
Erros Inatos do Metabolismo dos Aminoácidos/dietoterapia , Pesos e Medidas Corporais , Encefalopatias Metabólicas/dietoterapia , Alimentos Formulados , Glutaril-CoA Desidrogenase/deficiência , Lisina/administração & dosagem , Erros Inatos do Metabolismo dos Aminoácidos/sangue , Erros Inatos do Metabolismo dos Aminoácidos/metabolismo , Erros Inatos do Metabolismo dos Aminoácidos/fisiopatologia , Antropometria , Biomarcadores/análise , Biomarcadores/sangue , Encefalopatias Metabólicas/sangue , Encefalopatias Metabólicas/metabolismo , Encefalopatias Metabólicas/fisiopatologia , Carnitina/administração & dosagem , Criança , Pré-Escolar , Suplementos Nutricionais , Ingestão de Alimentos/fisiologia , Feminino , Seguimentos , Glutaril-CoA Desidrogenase/sangue , Glutaril-CoA Desidrogenase/metabolismo , Humanos , Lactente , Masculino , Monitorização Fisiológica/métodos
15.
Metab Brain Dis ; 26(3): 159-72, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21643764

RESUMO

Proline is an amino acid with an essential role for primary metabolism and physiologic functions. Hyperprolinemia results from the deficiency of specific enzymes for proline catabolism, leading to tissue accumulation of this amino acid. Hyperprolinemic patients can present neurological symptoms and brain abnormalities, whose aetiopathogenesis is poorly understood. This review addresses some of the findings obtained, mainly from animal studies, indicating that high proline levels may be associated to neuropathophysiology of some disorders. In this context, it has been suggested that energy metabolism deficit, Na(+),K(+)-ATPase, kinase creatine, oxidative stress, excitotoxicity, lipid content, as well as purinergic and cholinergic systems are involved in the effect of proline on brain damage and spatial memory deficit. The discussion focuses on the relatively low antioxidant defenses of the brain and the vulnerability of neural tissue to reactive species. This offers new perspectives for potential therapeutic strategies for this condition, which may include the early use of appropriate antioxidants as a novel adjuvant therapy, besides the usual treatment based on special diets poor in proline.


Assuntos
Antioxidantes , Encefalopatias Metabólicas , Metabolismo Energético/fisiologia , Radicais Livres/efeitos adversos , Transtornos da Memória , Estresse Oxidativo/fisiologia , Prolina , 1-Pirrolina-5-Carboxilato Desidrogenase/deficiência , Erros Inatos do Metabolismo dos Aminoácidos/metabolismo , Animais , Antioxidantes/metabolismo , Antioxidantes/uso terapêutico , Ácido Ascórbico/uso terapêutico , Encéfalo/metabolismo , Encefalopatias Metabólicas/induzido quimicamente , Encefalopatias Metabólicas/metabolismo , Creatina Quinase/metabolismo , Glicina/metabolismo , Glicina/urina , Atrofia Girata/metabolismo , Humanos , Transtornos da Memória/induzido quimicamente , Transtornos da Memória/metabolismo , Camundongos , Prolina/efeitos adversos , Prolina/metabolismo , Prolina Oxidase/deficiência , Prolina Oxidase/metabolismo , Ratos , Receptores Colinérgicos/metabolismo , Receptores Purinérgicos/metabolismo , Erros Inatos do Transporte Tubular Renal/metabolismo , ATPase Trocadora de Sódio-Potássio/metabolismo , Vitamina E/uso terapêutico
16.
J Neurol Neurosurg Psychiatry ; 82(8): 869-75, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21242286

RESUMO

OBJECTIVE: The authors examined whether (1)H-magnetic resonance spectroscopy (MRS) can identify damage to the centrum semiovale in the subacute phase after CO exposure. METHODS: Subjects comprised 29 adult patients who were treated with hyperbaric oxygenation within a range of 4-95 h (mean 18.7 h) after CO exposure. Subjects were classified into three groups according to clinical behaviours: Group A, patients with transit acute symptoms only; Group P, patients with persistent neurological symptoms; and Group D, patients with 'delayed neuropsychiatric sequelae' occurring after a lucid interval. MRS of bilateral centrum semiovale was performed 2 weeks after CO inhalation for all patients and 13 healthy volunteers. The mean ratios of choline-containing compounds/creatine ((mean)Cho/Cr) and N-acetylaspartate/Cr ((mean)NAA/Cr) for bilateral centrum semiovale were calculated and compared between the three CO groups and controls. Myelin basic protein (MBP) concentration in cerebrospinal fluid was examined at 2 weeks to evaluate the degree of demyelination in patients. RESULTS: MBP concentration was abnormal for almost all patients in Groups P and D, but was not abnormal for any Group A patients. The (mean)Cho/Cr ratios were significantly higher in Groups P and D than in Group A. No significant difference in (mean)NAA/Cr ratio was seen between the three pathological groups and controls. A significant correlation was identified between MBP and (mean)Cho/Cr ratio. CONCLUSIONS: These results suggest that the Cho/Cr ratio in the subacute phase after CO intoxication represents early demyelination in the centrum semiovale, and can predict chronic neurological symptoms.


Assuntos
Encefalopatias Metabólicas/diagnóstico , Encéfalo/fisiopatologia , Intoxicação por Monóxido de Carbono/fisiopatologia , Doenças Desmielinizantes/diagnóstico , Espectroscopia de Ressonância Magnética , Fibras Nervosas Mielinizadas/metabolismo , Encéfalo/metabolismo , Encefalopatias Metabólicas/etiologia , Encefalopatias Metabólicas/metabolismo , Intoxicação por Monóxido de Carbono/complicações , Intoxicação por Monóxido de Carbono/terapia , Colina/metabolismo , Creatina/metabolismo , Doenças Desmielinizantes/etiologia , Doenças Desmielinizantes/metabolismo , Feminino , Seguimentos , Humanos , Oxigenoterapia Hiperbárica , Masculino , Pessoa de Meia-Idade , Proteína Básica da Mielina/líquido cefalorraquidiano , Prótons , Fatores de Tempo
17.
Brain ; 134(Pt 1): 157-70, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20923787

RESUMO

Glutaric aciduria type I, an inherited deficiency of glutaryl-coenzyme A dehydrogenase localized in the final common catabolic pathway of L-lysine, L-hydroxylysine and L-tryptophan, leads to accumulation of neurotoxic glutaric and 3-hydroxyglutaric acid, as well as non-toxic glutarylcarnitine. Most untreated patients develop irreversible brain damage during infancy that can be prevented in the majority of cases if metabolic treatment with a low L-lysine diet and L-carnitine supplementation is started in the newborn period. The biochemical effect of this treatment remains uncertain, since cerebral concentrations of neurotoxic metabolites can only be determined by invasive techniques. Therefore, we studied the biochemical effect and mechanism of metabolic treatment in glutaryl-coenzyme A dehydrogenase-deficient mice, an animal model with complete loss of glutaryl-coenzyme A dehydrogenase activity, focusing on the tissue-specific changes of neurotoxic metabolites and key enzymes of L-lysine metabolism. Here, we demonstrate that low L-lysine diet, but not L-carnitine supplementation, lowered the concentration of glutaric acid in brain, liver, kidney and serum. L-carnitine supplementation restored the free L-carnitine pool and enhanced the formation of glutarylcarnitine. The effect of low L-lysine diet was amplified by add-on therapy with L-arginine, which we propose to result from competition with L-lysine at system y(+) of the blood-brain barrier and the mitochondrial L-ornithine carriers. L-lysine can be catabolized in the mitochondrial saccharopine or the peroxisomal pipecolate pathway. We detected high activity of mitochondrial 2-aminoadipate semialdehyde synthase, the rate-limiting enzyme of the saccharopine pathway, in the liver, whereas it was absent in the brain. Since we found activity of the subsequent enzymes of L-lysine oxidation, 2-aminoadipate semialdehyde dehydrogenase, 2-aminoadipate aminotransferase and 2-oxoglutarate dehydrogenase complex as well as peroxisomal pipecolic acid oxidase in brain tissue, we postulate that the pipecolate pathway is the major route of L-lysine degradation in the brain and the saccharopine pathway is the major route in the liver. Interestingly, treatment with clofibrate decreased cerebral and hepatic concentrations of glutaric acid in glutaryl-coenzyme A dehydrogenase-deficient mice. This finding opens new therapeutic perspectives such as pharmacological stimulation of alternative L-lysine oxidation in peroxisomes. In conclusion, this study gives insight into the discrepancies between cerebral and hepatic L-lysine metabolism, provides for the first time a biochemical proof of principle for metabolic treatment in glutaric aciduria type I and suggests that further optimization of treatment could be achieved by exploitation of competition between L-lysine and L-arginine at physiological barriers and enhancement of peroxisomal L-lysine oxidation and glutaric acid breakdown.


Assuntos
Encéfalo/metabolismo , Lisina/metabolismo , 2-Aminoadipato Transaminase/metabolismo , Ácido 2-Aminoadípico/análogos & derivados , Ácido 2-Aminoadípico/metabolismo , Erros Inatos do Metabolismo dos Aminoácidos/dietoterapia , Erros Inatos do Metabolismo dos Aminoácidos/metabolismo , Análise de Variância , Animais , Arginina/metabolismo , Arginina/uso terapêutico , Encefalopatias Metabólicas/dietoterapia , Encefalopatias Metabólicas/metabolismo , Carnitina/análogos & derivados , Carnitina/metabolismo , Carnitina/uso terapêutico , Catalase/metabolismo , Glutaril-CoA Desidrogenase/deficiência , Glutaril-CoA Desidrogenase/metabolismo , Ácidos Cetoglutáricos/metabolismo , Camundongos
18.
J Nutr Sci Vitaminol (Tokyo) ; 56(2): 87-97, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20495289

RESUMO

Diabetic encephalopathy is a severe complication in patients with long-term hyperglycemia. Oxidative stress is thought to be closely implicated in this disorder, so in this study, we examined whether grape seed proanthocyanidin extract (GSPE), a naturally occurring antioxidant derived from grape seeds, could reduce the injuries in the cerebral cortex of diabetic rats by modulating advanced glycation end products (AGEs)/the receptor for AGEs (RAGE)/nuclear factor-kappa B p65 (NF-kappaB p65) pathway, which is crucial in oxidative stress. Body weight and serum AGEs were tested; cerebral cortexes were isolated for morphological observations and the pyramidal cell layers were immunohistochemically stained for the detection of RAGE, NF-kappaB p65, intercellular adhesion molecule-1 (ICAM-1) and vascular cell adhesion molecule-1 (VCAM-1) as well. For RAGE and NF-kappaB p65, quantitative reverse transcriptase coupled to polymerase chain reaction (RT-PCR) was employed for determination of mRNA levels, and western blot was used to detect protein expression. Our results showed that long term hyperglycemia in diabetic rats caused the degeneration of neurons and the up-regulation of serum AGEs, and also the up-regulation of RAGE, NF-kappaB p65, VCAM-1 and ICAM-1 in the brain. We found that GSPE treatment improved the pathological changes of diabetic rats by modulating the AGEs/RAGE/NF-kappaB p65 pathway. This study enables us to further understand the key role that the AGEs/RAGE/NF-kappaB pathway plays in the pathogenesis of diabetic encephalopathy, and confirms that GSPE might be a therapeutical means to the prevention and treatment of this disorder.


Assuntos
Encefalopatias Metabólicas/tratamento farmacológico , Córtex Cerebral/efeitos dos fármacos , Complicações do Diabetes/tratamento farmacológico , Diabetes Mellitus Experimental/metabolismo , Produtos Finais de Glicação Avançada/metabolismo , Extrato de Sementes de Uva/uso terapêutico , NF-kappa B/metabolismo , Proantocianidinas/uso terapêutico , Animais , Antioxidantes/uso terapêutico , Western Blotting/métodos , Peso Corporal/efeitos dos fármacos , Encefalopatias Metabólicas/metabolismo , Diabetes Mellitus Experimental/sangue , Diabetes Mellitus Experimental/induzido quimicamente , Diabetes Mellitus Experimental/tratamento farmacológico , Modelos Animais de Doenças , Produtos Finais de Glicação Avançada/sangue , Molécula 1 de Adesão Intercelular/efeitos dos fármacos , Molécula 1 de Adesão Intercelular/metabolismo , Masculino , NF-kappa B/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Ratos , Ratos Wistar , Receptor para Produtos Finais de Glicação Avançada , Receptores Imunológicos/efeitos dos fármacos , Receptores Imunológicos/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos , Estreptozocina , Molécula 1 de Adesão de Célula Vascular/efeitos dos fármacos , Molécula 1 de Adesão de Célula Vascular/metabolismo
19.
Metab Brain Dis ; 25(2): 227-34, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20424902

RESUMO

Acute hyperglycaemia impairs cognitive function. It is however not known, whether different brain regions are equally exposed to glucose during acute hyperglycemia or whether the brain is able to adjust its glucose uptake or metabolism in response to blood glucose fluctuation. We studied the effect of acute hyperglycaemia on the brain glucose concentration in seven men with type 1 diabetes with daily glucose fluctuations of 11 +/- 3 mmol/l, and in eleven age-matched non-diabetic men. Glucose was quantified with proton magnetic resonance spectroscopy in three different brain regions at baseline (fasting glycaemia) and twice during a 2 h hyperglycaemic clamp with plasma glucose increase of 12 mmol/l. The increase in brain glucose during acute hyperglycaemia in the non-diabetic group was: cortex (2.7 +/- 0.9 mmol/l) > thalamus (2.3 +/- 0.7 mmol/l) > white matter (1.7 +/- 0.7 mmol/l, P = 0.021 vs. cortex) and in the diabetic group: cortex (2.0 +/- 0.7 mmol/l) > white matter (1.3 +/- 0.7 mmol/l) > thalamus (1.1 +/- 0.4 mmol/l, P = 0.010 vs. cortex). In the diabetic group, the glucose increase in the thalamus was attenuated compared to the non-diabetic participants (P = 0.011). In conclusion, the increase of glucose during acute hyperglycaemia seems to be dependent on the brain tissue type. The high exposure of cortex to excess glucose and the altered glucose uptake or metabolism in the thalamus may thus contribute to hyperglycaemia related cognitive dysfunction.


Assuntos
Encefalopatias Metabólicas/metabolismo , Diabetes Mellitus Tipo 1/metabolismo , Glucose/metabolismo , Hiperglicemia/metabolismo , Tálamo/metabolismo , Doença Aguda , Adulto , Encefalopatias Metabólicas/sangue , Encefalopatias Metabólicas/complicações , Córtex Cerebral/metabolismo , Transtornos Cognitivos/metabolismo , Diabetes Mellitus Tipo 1/sangue , Diabetes Mellitus Tipo 1/complicações , Humanos , Hiperglicemia/sangue , Hiperglicemia/complicações , Espectroscopia de Ressonância Magnética , Masculino , Adulto Jovem
20.
Brain ; 132(Pt 12): 3285-97, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19952056

RESUMO

Mitochondrial dysfunction hypothetically contributes to neuronal degeneration in patients with Parkinson's disease. While several in vitro data exist, the measurement of cerebral mitochondrial dysfunction in living patients with Parkinson's disease is challenging. Anatomical magnetic resonance imaging combined with phosphorus and proton magnetic resonance spectroscopic imaging provides information about the functional integrity of mitochondria in specific brain areas. We measured partial volume corrected concentrations of low-energy metabolites and high-energy phosphates with sufficient resolution to focus on pathology related target areas in Parkinson's disease. Combined phosphorus and proton magnetic resonance spectroscopic imaging in the mesostriatal region was performed in 16 early and 13 advanced patients with Parkinson's disease and compared to 19 age-matched controls at 3 Tesla. In the putamen and midbrain of both Parkinson's disease groups, we found a bilateral reduction of high-energy phosphates such as adenosine triphophosphate and phosphocreatine as final acceptors of energy from mitochondrial oxidative phosphorylation. In contrast, low-energy metabolites such as adenosine diphophosphate and inorganic phosphate were within normal ranges. These results provide strong in vivo evidence that mitochondrial dysfunction of mesostriatal neurons is a central and persistent phenomenon in the pathogenesis cascade of Parkinson's disease which occurs early in the course of the disease.


Assuntos
Encefalopatias Metabólicas/metabolismo , Encéfalo/metabolismo , Metabolismo Energético/fisiologia , Mitocôndrias/metabolismo , Doenças Mitocondriais/metabolismo , Doença de Parkinson/metabolismo , Trifosfato de Adenosina/análise , Trifosfato de Adenosina/metabolismo , Idoso , Biomarcadores/análise , Biomarcadores/metabolismo , Encéfalo/fisiopatologia , Química Encefálica/fisiologia , Encefalopatias Metabólicas/diagnóstico , Encefalopatias Metabólicas/fisiopatologia , Progressão da Doença , Feminino , Humanos , Espectroscopia de Ressonância Magnética/métodos , Masculino , Pessoa de Meia-Idade , Doenças Mitocondriais/diagnóstico , Doenças Mitocondriais/fisiopatologia , Fosforilação Oxidativa , Doença de Parkinson/diagnóstico , Doença de Parkinson/fisiopatologia , Fosfocreatina/análise , Fosfocreatina/metabolismo , Fósforo/metabolismo , Valor Preditivo dos Testes , Prótons , Putamen/metabolismo , Putamen/fisiopatologia , Substância Negra/metabolismo , Substância Negra/fisiopatologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA